Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Stem Cells Dev ; 23(5): 530-40, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24147599

RESUMO

The goal of this study was to determine whether a pro-survival cocktail (PSC, consisting of IGF-1, Bcl-XL, and Caspase-I Inhibitor) and long-term hypoxia (LTH) enhance survival and functional properties of bone marrow-derived stromal stem cells (BMSCs), in response to stress conditions. PSC-treated cells retained BMSC surface markers and protected cells from apoptosis under serum starvation and ischemic (1% O2 and 100 µM H2O2) conditions. LTH promoted osteogenesis, while suppressing adipogenesis. LTH alone did not result in an improvement in the apoptosis rate; however, PSC conferred significant protection regardless of the oxygenation status. One of the possible mechanisms of PSC protection was due to the elevated phospho-AKT in treated groups. PSC treatment or LTH did not alter migration toward stem cell-derived factor-1 alpha (SDF-1α) or fetal bovine serum, nor did they enhance cell motility during wound healing. There was no difference in the secreted cytokine profiles of BMSCs treated with PSC after stress when grown in normoxic or LTH. However, LTH did upregulate the vascular endothelial growth factor, hepatocyte growth factor, and SDF-1α, while it downregulated other anti- and proinflammatory cytokines and chemokines. We also observed a high degree of interdonor BMSC variability in response to pretreatment with PSC and LTH, confounding the functional results, underscoring the observation that not all donor-derived BMSCs will respond similarly.


Assuntos
Adipogenia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Hipóxia Celular , Osteogênese/efeitos dos fármacos , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Inibidores de Caspase/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Humanos , Fator de Crescimento Insulin-Like I/administração & dosagem , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Proteína bcl-X/administração & dosagem
2.
Stem Cells ; 30(6): 1216-27, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22593018

RESUMO

Bone marrow stromal cells (BMSCs) have shown significant promise in the treatment of disease, but their therapeutic efficacy is often limited by inefficient homing of systemically administered cells, which results in low number of cells accumulating at sites of pathology. BMSC home to areas of inflammation where local expression of integrins and chemokine gradients is present. We demonstrated that nondestructive pulsed focused ultrasound (pFUS) exposures that emphasize the mechanical effects of ultrasound-tissue interactions induced local and transient elevations of chemoattractants (i.e., cytokines, integrins, and growth factors) in the murine kidney. pFUS-induced upregulation of cytokines occurred through approximately 1 day post-treatment and returned to contralateral kidney levels by day 3. This window of significant increases in cytokine expression was accompanied by local increases of other trophic factors and integrins that have been shown to promote BMSC homing. When BMSCs were intravenously administered following pFUS treatment to a single kidney, enhanced homing, permeability, and retention of BMSC was observed in the treated kidney versus the contralateral kidney. Histological analysis revealed up to eight times more BMSC in the peritubular regions of the treated kidneys on days 1 and 3 post-treatment. Furthermore, cytokine levels in pFUS-treated kidneys following BMSC administration were found to be similar to controls, suggesting modulation of cytokine levels by BMSC. pFUS could potentially improve cell-based therapies as a noninvasive modality to target homing by establishing local chemoattractant gradients and increasing expression of integrins to enhance tropism of cells toward treated tissues.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/diagnóstico por imagem , Transplante de Medula Óssea/métodos , Rim/citologia , Rim/diagnóstico por imagem , Células Estromais/transplante , Ultrassom/métodos , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/diagnóstico por imagem , Técnicas de Cultura de Células , Citocinas/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células Estromais/citologia , Ultrassonografia
3.
Nat Med ; 18(3): 463-7, 2012 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-22366951

RESUMO

We report on a new straightforward magnetic cell-labeling approach that combines three US Food and Drug Administration (FDA)-approved drugs--ferumoxytol, heparin and protamine--in serum-free medium to form self-assembling nanocomplexes that effectively label cells for in vivo magnetic resonance imaging (MRI). We observed that the ferumoxytol-heparin-protamine (HPF) nanocomplexes were stable in serum-free cell culture medium. HPF nanocomplexes show a threefold increase in T2 relaxivity compared to ferumoxytol. Electron microscopy showed internalized HPF in endosomes, which we confirmed by Prussian blue staining of labeled cells. There was no long-term effect or toxicity on cellular physiology or function of HPF-labeled hematopoietic stem cells, bone marrow stromal cells, neural stem cells or T cells when compared to controls. In vivo MRI detected 1,000 HPF-labeled cells implanted in rat brains. This HPF labeling method should facilitate the monitoring by MRI of infused or implanted cells in clinical trials.


Assuntos
Encéfalo/citologia , Rastreamento de Células/métodos , Óxido Ferroso-Férrico/química , Heparina/química , Nanopartículas de Magnetita/administração & dosagem , Nanopartículas de Magnetita/efeitos adversos , Nanopartículas de Magnetita/química , Protaminas/química , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/citologia , Encéfalo/diagnóstico por imagem , Encéfalo/ultraestrutura , Diferenciação Celular/efeitos dos fármacos , Ensaios Clínicos como Assunto , Meios de Cultura Livres de Soro , Endossomos/ultraestrutura , Óxido Ferroso-Férrico/administração & dosagem , Óxido Ferroso-Férrico/efeitos adversos , Células-Tronco Hematopoéticas/citologia , Heparina/administração & dosagem , Heparina/efeitos adversos , Humanos , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/ultraestrutura , Masculino , Células-Tronco Mesenquimais/citologia , Microscopia Eletrônica , Neurônios/citologia , Protaminas/administração & dosagem , Protaminas/efeitos adversos , Radiografia , Ratos , Transplante de Células-Tronco , Células Estromais/citologia , Linfócitos T/citologia
4.
Mol Imaging Biol ; 14(3): 325-35, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21614680

RESUMO

PURPOSE: The sigma-2 (σ(2)) receptor is a potential biomarker of proliferative status of solid tumors. Specific synthetic probes using N-substituted-9-azabicyclo [3.3.1]nonan-3α-yl carbamate analogs have been designed and implemented for experimental cancer diagnosis and therapy. PROCEDURES: We employed the fluorescently labeled σ(2) receptor probe, SW120, to evaluate σ(2) receptor expression in human stem cells (SC), including: bone marrow stromal, neural progenitor, amniotic fluid, hematopoetic, and embryonic stem cells. We concurrently evaluated the intensity of SW120 and 5-ethynyl-2'-deoxyuridine (EdU) relative to passage number and multi-potency. RESULTS: We substantiated significantly higher σ(2) receptor density among proliferating SC relative to lineage-restricted cell types. Additionally, cellular internalization of the σ(2) receptor in SC was consistent with receptor-mediated endocytosis and confocal microscopy indicated SW120 specific co-localization with a fluorescent marker of lysosomes in all SC imaged. CONCLUSION: These results suggest that σ(2) receptors may serve to monitor stem cell differentiation in future experimental studies.


Assuntos
Diferenciação Celular/fisiologia , Corantes Fluorescentes/metabolismo , Receptores sigma/metabolismo , Células-Tronco/citologia , Líquido Amniótico/citologia , Animais , Apoptose , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/química , Biomarcadores Tumorais/metabolismo , Células da Medula Óssea/citologia , Linhagem Celular Tumoral , Corantes Fluorescentes/análise , Humanos , Fenótipo , Ratos , Receptores sigma/análise , Receptores sigma/química , Espectrometria de Fluorescência , Células-Tronco/química , Células-Tronco/metabolismo
5.
PLoS One ; 6(9): e24730, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21931834

RESUMO

Continuous focused ultrasound (cFUS) has been widely used for thermal ablation of tissues, relying on continuous exposures to generate temperatures necessary to induce coagulative necrosis. Pulsed FUS (pFUS) employs non-continuous exposures that lower the rate of energy deposition and allow cooling to occur between pulses, thereby minimizing thermal effects and emphasizing effects created by non-thermal mechanisms of FUS (i.e., acoustic radiation forces and acoustic cavitation). pFUS has shown promise for a variety of applications including drug and nanoparticle delivery; however, little is understood about the effects these exposures have on tissue, especially with regard to cellular pro-homing factors (growth factors, cytokines, and cell adhesion molecules). We examined changes in murine hamstring muscle following pFUS or cFUS and demonstrate that pFUS, unlike cFUS, has little effect on the histological integrity of muscle and does not induce cell death. Infiltration of macrophages was observed 3 and 8 days following pFUS or cFUS exposures. pFUS increased expression of several cytokines (e.g., IL-1α, IL-1ß, TNFα, INFγ, MIP-1α, MCP-1, and GMCSF) creating a local cytokine gradient on days 0 and 1 post-pFUS that returns to baseline levels by day 3 post-pFUS. pFUS exposures induced upregulation of other signaling molecules (e.g., VEGF, FGF, PlGF, HGF, and SDF-1α) and cell adhesion molecules (e.g., ICAM-1 and VCAM-1) on muscle vasculature. The observed molecular changes in muscle following pFUS may be utilized to target cellular therapies by increasing homing to areas of pathology.


Assuntos
Ablação por Ultrassom Focalizado de Alta Intensidade , Animais , Apoptose/fisiologia , Moléculas de Adesão Celular/metabolismo , Quimiocina CCL3/metabolismo , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-1/metabolismo , Interleucina-1beta/metabolismo , Macrófagos , Imageamento por Ressonância Magnética , Camundongos , Músculo Esquelético/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
6.
J Transl Med ; 9: 145, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21871109

RESUMO

BACKGROUND AND PURPOSE: Both endothelial progenitor cells (EPC) and markers of neuroinflammation are candidate biomarkers for stroke severity and outcome prediction. A relationship between EPC and neuroinflammatory markers in early stroke is not fully elucidated. The objectives were to investigate correlations between EPC and neuroinflammation markers (adhesion molecules ICAM-1, VCAM-1, E-selectin, tumor necrosis factor (TNF)-α, interleukin (IL)-6, endothelin (ET)-1, markers of tissue injury (matrix metalloproteinases (MMP)-9 and tissue inhibitor of matrix metalloproteinases (TIMP)-1) in early stroke patients. METHODS: We prospectively recruited symptomatic patients with ischemic cerebrovascular disease. We assessed stroke severity by using of acute (diffusion-weighted imaging (DWI) and final lesion volumes (fluid attenuated inversion recovery (FLAIR). We measured serum soluble ICAM-1, VCAM-1, E-selectin, MMP-9, TIMP-1 and plasma TNF-α, IL-6, ET-1 by ELISA, and quantified EPC in mononuclear fraction of peripheral blood on days 1 and 3 in 17 patients (mean(SD) age 62(14), with admission National Institutes of Health Stroke Scale (NIHSS) 10(8)) selected from 175 patients with imaging confirmed ischemic stroke. Non-parametric statistics, univariate and multivariate analysis were used. RESULTS: Only ICAM-1 inversely correlated with EPC subset CD133+CD34+ on day 1 (Spearman r = -0.6, p < 0.01) and on day 3 (r = -0.967, p < 0.001). This correlation remained significant after adjustment for age and NIHSS (beta -0.992, p < 0.004), for glucose and systolic blood pressure (beta -0.86, p < 0.005), and for white blood cells and hematocrit (beta -1.057, p < 0.0001) on day 3. MMP-9 (r = 0.509, p < 0.04) and MMP-9/TIMP-1 (r = 0.59, p < 0.013) on day 1 correlated with acute lesion volume. Both IL-6 (r = 0.624, p < 0.01) and MMP-9/TIMP-1 (r = 0.56, p < 0.02) correlated with admission NIHSS. CONCLUSION: Our study showed that high ICAM-1 is associated with low CD133+CD34+subset of EPC. Biomarkers of neuroinflammation may predict tissue injury and stroke severity in early ischemia.


Assuntos
Antígenos CD34/metabolismo , Antígenos CD/metabolismo , Isquemia Encefálica/sangue , Movimento Celular , Glicoproteínas/metabolismo , Molécula 1 de Adesão Intercelular/sangue , Peptídeos/metabolismo , Células-Tronco/metabolismo , Acidente Vascular Cerebral/sangue , Antígeno AC133 , Biomarcadores/metabolismo , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Demografia , Células Endoteliais/metabolismo , Feminino , Humanos , Masculino , Metaloproteinase 9 da Matriz/sangue , Pessoa de Meia-Idade , Fatores de Risco , Solubilidade , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia , Fatores de Tempo , Inibidor Tecidual de Metaloproteinase-1/sangue
7.
PLoS One ; 5(7): e11462, 2010 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-20628641

RESUMO

Superparamagnetic iron oxide nanoparticles (SPION) are increasingly used to label human bone marrow stromal cells (BMSCs, also called "mesenchymal stem cells") to monitor their fate by in vivo MRI, and by histology after Prussian blue (PB) staining. SPION-labeling appears to be safe as assessed by in vitro differentiation of BMSCs, however, we chose to resolve the question of the effect of labeling on maintaining the "stemness" of cells within the BMSC population in vivo. Assays performed include colony forming efficiency, CD146 expression, gene expression profiling, and the "gold standard" of evaluating bone and myelosupportive stroma formation in vivo in immuncompromised recipients. SPION-labeling did not alter these assays. Comparable abundant bone with adjoining host hematopoietic cells were seen in cohorts of mice that were implanted with SPION-labeled or unlabeled BMSCs. PB+ adipocytes were noted, demonstrating their donor origin, as well as PB+ pericytes, indicative of self-renewal of the stem cell in the BMSC population. This study confirms that SPION labeling does not alter the differentiation potential of the subset of stem cells within BMSCs.


Assuntos
Células da Medula Óssea/citologia , Óxido Ferroso-Férrico/química , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/citologia , Nanopartículas/química , Animais , Antígeno CD146/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Óxido Ferroso-Férrico/efeitos adversos , Perfilação da Expressão Gênica , Humanos , Imageamento por Ressonância Magnética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Nanopartículas/efeitos adversos , Análise de Sequência com Séries de Oligonucleotídeos
8.
PLoS One ; 4(8): e6712, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19696933

RESUMO

Intracellular labels such as dextran coated superparamagnetic iron oxide nanoparticles (SPION), bromodeoxyuridine (BrdU) or green fluorescent protein (GFP) are frequently used to study the fate of transplanted cells by in vivo magnetic resonance imaging or fluorescent microscopy. Bystander uptake of labeled cells by resident tissue macrophages (TM) can confound the interpretation of the presence of intracellular labels especially during direct implantation of cells, which can result in more than 70% cell death. In this study we determined the percentages of TM that took up SPION, BrdU or GFP from labeled bone marrow stromal cells (BMSCs) that were placed into areas of angiogenesis and inflammation in a mouse model known as Matrigel plaque perfusion assay. Cells recovered from digested plaques at various time points were analyzed by fluorescence microscopy and flow cytometry. The analysis of harvested plaques revealed 5% of BrdU(+), 5-10% of GFP(+) and 5-15% of dextran(+) macrophages. The transfer of the label was not dependent on cell dose or viability. Collectively, this study suggests that care should be taken to validate donor origin of cells using an independent marker by histology and to assess transplanted cells for TM markers prior to drawing conclusions about the in vivo behavior of transplanted cells.


Assuntos
Células da Medula Óssea/citologia , Macrófagos/citologia , Células Estromais/citologia , Animais , Bromodesoxiuridina/metabolismo , Linhagem da Célula , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Humanos , Imageamento por Ressonância Magnética , Camundongos , Microscopia de Fluorescência
9.
Stem Cells ; 26(5): 1366-75, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18276802

RESUMO

There is increasing interest in using exogenous labels such as bromodeoxyuridine (BrdU) or superparamagnetic iron oxide nanoparticles (SPION) to label cells to identify transplanted cells and monitor their migration by fluorescent microscopy or in vivo magnetic resonance imaging (MRI), respectively. Direct implantation of cells into target tissue can result in >80% cell death due to trauma or apoptosis. Bystander uptake of labeled cells by activated macrophages (AM) can confound the interpretation of results. This study investigated the frequency of BrdU or SPION uptake by AM using the Boyden chamber model of inflammation. SPION/BrdU-labeled bone marrow stromal cells or HeLa cells, AM, and mouse fibroblasts (MF) or human fibroblasts (HF) were mixed in various ratios in Matrigel in the upper chamber and incubated for up to 96 hours. The AM were chemotactically induced to migrate to the lower chamber. Fluorescence-activated cell sorting analysis of AM from lower and upper chambers, in the presence of either MF or HF using anti-CD68, anti-BrdU, anti-dextran antibodies, revealed 10%-20% dextran-positive or 10% BrdU-positive AM after 96 hours of incubation. Transfer of iron to AM accounted for <10% of the total iron in labeled cells. The uptake of BrdU and SPION was dependent on the ratio of labeled cells to inflammatory cells and microenvironmental conditions. Direct implantation of BrdU/SPION-labeled cells into target tissue can result in uptake of label by AM; therefore, care should be taken to validate by histology transplanted cells for bystander cell markers and correlation with MRI results.


Assuntos
Bromodesoxiuridina/metabolismo , Terapia Baseada em Transplante de Células e Tecidos , Ferro/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Nanopartículas , Óxidos/metabolismo , Células-Tronco/metabolismo , Animais , Apoptose , Sobrevivência Celular , Células Cultivadas , Dextranos , Óxido Ferroso-Férrico , Imunofluorescência , Humanos , Macrófagos/citologia , Nanopartículas de Magnetita , Camundongos , Protaminas/metabolismo , Reprodutibilidade dos Testes , Coloração e Rotulagem
10.
Stem Cells ; 25(10): 2524-33, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17628016

RESUMO

Brain tumors can arise following deregulation of signaling pathways normally activated during brain development and may derive from neural stem cells. Given the requirement for Hedgehog in non-neoplastic stem cells, we investigated whether Hedgehog blockade could target the stem-like population in glioblastoma multiforme (GBM). We found that Gli1, a key Hedgehog pathway target, was highly expressed in 5 of 19 primary GBM and in 4 of 7 GBM cell lines. Shh ligand was expressed in some primary tumors, and in GBM-derived neurospheres, suggesting a potential mechanism for pathway activation. Hedgehog pathway blockade by cyclopamine caused a 40%-60% reduction in growth of adherent glioma lines highly expressing Gli1 but not in those lacking evidence of pathway activity. When GBM-derived neurospheres were treated with cyclopamine and then dissociated and seeded in media lacking the inhibitor, no new neurospheres formed, suggesting that the clonogenic cancer stem cells had been depleted. Consistent with this hypothesis, the stem-like fraction in gliomas marked by both aldehyde dehydrogenase activity and Hoechst dye excretion (side population) was significantly reduced or eliminated by cyclopamine. In contrast, we found that radiation treatment of our GBM neurospheres increased the percentage of these stem-like cells, suggesting that this standard therapy preferentially targets better-differentiated neoplastic cells. Most importantly, viable GBM cells injected intracranially following Hedgehog blockade were no longer able to form tumors in athymic mice, indicating that a cancer stem cell population critical for ongoing growth had been removed. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Glioblastoma/patologia , Proteínas Hedgehog/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Células-Tronco Neoplásicas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Alcaloides de Veratrum/farmacologia , Animais , Linhagem Celular Tumoral/efeitos dos fármacos , Etanol/farmacologia , Raios gama , Proteínas Hedgehog/biossíntese , Proteínas Hedgehog/genética , Proteínas Hedgehog/fisiologia , Humanos , Camundongos , Camundongos Nus , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Transplante de Neoplasias , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/efeitos da radiação , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Proteína GLI1 em Dedos de Zinco
11.
Am J Pathol ; 170(1): 347-55, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17200206

RESUMO

Activation of the Hedgehog (Hh) pathway has been identified in several cancers, including medulloblastoma, but the mechanisms by which this pathway affects tumor survival and growth are incompletely understood. We investigated whether Hedgehog might promote survival of medulloblastoma cells via up-regulation of BclII. We found that mRNA levels of the Hedgehog pathway effector Gli1 were significantly associated with BclII expression in medulloblastoma and that Gli1 and BclII are both present in regions of decreased apoptosis in nodular medulloblastoma. Transient overexpression of Gli1 and Gli2 in medulloblastoma cultures induced a BclII transcriptional reporter and increased BclII protein levels, whereas stable overexpression of Gli1 was associated with increased BclII mRNA. The Hedgehog antagonist cyclopamine blocked expression of the Hh pathway targets PTCH1 and Gli1, lowered BclII levels, and increased apoptosis in DAOY and UW228 medulloblastoma cells. Apoptotic induction caused by cyclopamine could be rescued in part by enforced expression of Gli1 or BclII. Hh pathway blockade also sensitized medulloblastoma to the effects of the proapoptotic agent lovastatin. These data demonstrate that BclII is an important mediator of Hh activity in medulloblastoma and suggest new strategies for combined chemotherapeutic regimens.


Assuntos
Neoplasias Cerebelares/metabolismo , Proteínas Hedgehog/metabolismo , Meduloblastoma/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Neoplasias Cerebelares/patologia , Humanos , Meduloblastoma/patologia , Fatores de Transcrição/metabolismo , Regulação para Cima , Alcaloides de Veratrum/farmacologia , Proteína GLI1 em Dedos de Zinco
12.
Cancer Res ; 66(2): 673-81, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16423996

RESUMO

Both anaplasia and increased c-myc gene expression have been shown to be negative prognostic indicators for survival in medulloblastoma patients. myc gene amplification has been identified in many large cell/anaplastic medulloblastoma, but no causative link between c-myc and anaplastic changes has been established. To address this, we stably overexpressed c-myc in two medulloblastoma cell lines, DAOY and UW228, and examined the changes in growth characteristics. When analyzed in vitro, cell lines with increased levels of c-myc had higher rates of growth and apoptosis as well as significantly improved ability to form colonies in soft agar compared with control. When injected s.c. into nu/nu mice, flank xenograft tumors with high levels of c-myc in DAOY cell line background were 75% larger than those derived from control. Overexpression of c-myc was required for tumor formation by UW228 cells. Most remarkably, the histopathology of the Myc tumors was severely anaplastic, with large areas of necrosis/apoptosis, increased nuclear size, and macronucleoli. Indices of proliferation and apoptosis were also significantly higher in Myc xenografts. Thus, c-myc seems to play a causal role in inducing anaplasia in medulloblastoma. Because anaplastic changes are often observed in recurrent medulloblastoma, we propose that c-myc dysregulation is involved in the progression of these malignant embryonal neoplasms.


Assuntos
Neoplasias Cerebelares/genética , Meduloblastoma/genética , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/fisiologia , Anaplasia , Animais , Apoptose , Proliferação de Células , Neoplasias Cerebelares/patologia , Humanos , Meduloblastoma/patologia , Camundongos , Necrose , Transplante Heterólogo , Células Tumorais Cultivadas
13.
BMC Cancer ; 5: 19, 2005 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-15717928

RESUMO

BACKGROUND: p53 mutations are relatively uncommon in medulloblastoma, but abnormalities in this cell cycle pathway have been associated with anaplasia and worse clinical outcomes. We correlated p53 protein expression with pathological subtype and clinical outcome in 75 embryonal brain tumors. The presence of JC virus, which results in p53 protein accumulation, was also examined. METHODS: p53 protein levels were evaluated semi-quantitatively in 64 medulloblastomas, 3 atypical teratoid rhabdoid tumors (ATRT), and 8 supratentorial primitive neuroectodermal tumors (sPNET) using immunohistochemistry. JC viral sequences were analyzed in DNA extracted from 33 frozen medulloblastoma and PNET samples using quantitative polymerase chain reaction. RESULTS: p53 expression was detected in 18% of non-anaplastic medulloblastomas, 45% of anaplastic medulloblastomas, 67% of ATRT, and 88% of sPNET. The increased p53 immunoreactivity in anaplastic medulloblastoma, ATRT, and sPNET was statistically significant. Log rank analysis of clinical outcome revealed significantly shorter survival in patients with p53 immunopositive embryonal tumors. No JC virus was identified in the embryonal brain tumor samples, while an endogenous human retrovirus (ERV-3) was readily detected. CONCLUSION: Immunoreactivity for p53 protein is more common in anaplastic medulloblastomas, ATRT and sPNET than in non-anaplastic tumors, and is associated with worse clinical outcomes. However, JC virus infection is not responsible for increased levels of p53 protein.


Assuntos
Vírus JC/isolamento & purificação , Meduloblastoma/metabolismo , Meduloblastoma/virologia , Neoplasias Supratentoriais/metabolismo , Neoplasias Supratentoriais/virologia , Proteína Supressora de Tumor p53/metabolismo , Adolescente , Adulto , Anaplasia , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Neoplasias Cerebelares/virologia , Criança , Pré-Escolar , Humanos , Imuno-Histoquímica , Lactente , Meduloblastoma/patologia , Pessoa de Meia-Idade , Tumores Neuroectodérmicos Primitivos/metabolismo , Tumores Neuroectodérmicos Primitivos/patologia , Tumores Neuroectodérmicos Primitivos/virologia , Neoplasias Supratentoriais/patologia , Teratoma/metabolismo , Teratoma/patologia , Teratoma/virologia
14.
Invest Ophthalmol Vis Sci ; 43(6): 2001-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12037011

RESUMO

PURPOSE: Platelet-derived growth factor (PDGF) has been implicated in vascular proliferative retinopathies, such as diabetic retinopathy, and in nonvascular retinopathies, such as proliferative vitreoretinopathy. Traction retinal detachment is a central feature of both types of disease. Hemizygous rhodopsin promoter/PDGF-B (rho/PDGF-B) transgenic mice exhibit proliferation of vascular cells, glia, and retinal pigmented epithelial (RPE) cells, resulting in traction retinal detachment. Hemizygous rho/PDGF-A transgenic mice show mild proliferation of glial cells and no traction retinal detachments. This study was undertaken to determine whether higher levels of endogenously produced PDGF-A in the retinas of mice result in retinal detachment. METHODS: To achieve high-level expression of PDGF-A in the retina, homozygous rho/PDGF-A (rho/PDGF-AA) mice were generated. The phenotype of these mice was compared with that of homozygous rho/PDGF-B (rho/PDGF-BB) mice and double hemizygous rho/PDGF-B-rho/PDGF-A (rho/PDGF-AB) mice. RESULTS: Rho/PDGF-BB and rho/PDGF-AB mice showed a phenotype similar to that previously described in rho/PDGF-B mice. There was extensive proliferation of glial and vascular cells, resulting in fibrovascular membranes that detached the retina. PDGF-AA mice showed extensive proliferation of glial cells and traction retinal detachment. CONCLUSIONS: High retinal expression of PDGF-A results in extensive proliferation of glial cells and traction retinal detachment without vascular cell involvement, similar to proliferative vitreoretinopathy in humans. High retinal expression of PDGF-B results in traction retinal detachment from proliferation of both vascular and nonvascular cells, similar to diabetic retinopathy in humans.


Assuntos
Retinopatia Diabética/metabolismo , Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Retina/metabolismo , Descolamento Retiniano/metabolismo , Vitreorretinopatia Proliferativa/metabolismo , Animais , Divisão Celular , Retinopatia Diabética/patologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroglia/metabolismo , Neuroglia/patologia , Retina/patologia , Descolamento Retiniano/patologia , Transgenes , Vitreorretinopatia Proliferativa/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA